WO2017037292A1 - Combination of a dhodh inhibitor and a her inhibitor for use in the treatment of cancer - Google Patents

Combination of a dhodh inhibitor and a her inhibitor for use in the treatment of cancer Download PDF

Info

Publication number
WO2017037292A1
WO2017037292A1 PCT/EP2016/070874 EP2016070874W WO2017037292A1 WO 2017037292 A1 WO2017037292 A1 WO 2017037292A1 EP 2016070874 W EP2016070874 W EP 2016070874W WO 2017037292 A1 WO2017037292 A1 WO 2017037292A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
group
inhibitor
heterocyclyl
alkyl
Prior art date
Application number
PCT/EP2016/070874
Other languages
English (en)
French (fr)
Inventor
Bertil Lindmark
Mark Thomas Mchale
Lisa OOI
Original Assignee
Aslan Pharmaceuticals Pte Limited
Sterling Ip
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GBGB1515718.3A external-priority patent/GB201515718D0/en
Priority claimed from GBGB1515716.7A external-priority patent/GB201515716D0/en
Priority claimed from GBGB1515712.6A external-priority patent/GB201515712D0/en
Priority claimed from GBGB1515714.2A external-priority patent/GB201515714D0/en
Priority claimed from GBGB1608660.5A external-priority patent/GB201608660D0/en
Application filed by Aslan Pharmaceuticals Pte Limited, Sterling Ip filed Critical Aslan Pharmaceuticals Pte Limited
Publication of WO2017037292A1 publication Critical patent/WO2017037292A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • A61K31/51Thiamines, e.g. vitamin B1
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/555Heterocyclic compounds containing heavy metals, e.g. hemin, hematin, melarsoprol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/86Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 4
    • C07D239/94Nitrogen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/282Platinum compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine

Definitions

  • the present disclosure relates to a method of treating cancer with a combination therapy, and a combination therapy for use in the treatment of cancer, in particular a cancer disclosed herein.
  • Tumor biopsies taken before and after treatment were analysed using immunohistochemistry. Signs of clinical activity included downregulation of signalling pathways responsible for cell proliferation, and a reduction in cell survival and reduced cell proliferation in gastric tumors that were either co-expressing EGFR and HER2 or that were HER2 amplified.
  • the HER2 specific monoclonal antibody herceptin is being developed as an antibody drug conjugate with a toxin payload to increase efficacy (ado-trastuzumab-emtansine, a so-called second generation cancer treatment).
  • HER inhibitors such as HER2 inhibitors or pan-HER inhibitors.
  • Cancer mechanisms generating resistance to treatment are complex and multifaceted. Thus cancers have a variety of routes for protecting themselves against the immune system and also cancer treatments.
  • HER2/neu Some work done on HER2/neu suggests that amplification or overexpression of this gene can render cancer cells resistant to apoptosis (programmed cell death) and promote cancer cell growth.
  • p53 is crucial in regulating a cell growth and apoptosis. Whilst not wishing to be bound by theory the present inventors believe, HER2/neu mediated resistance in cancer cells may be a result of activation of Akt, which enhances MDM2-mediated ubiquitination and degradation of p53.
  • the present disclosure provides an alternative combination for the treatment of cancer, which may fight cancer employing different combinations of mechanisms to those employed in known therapies.
  • the presently disclosed therapy may increase the repertoire of cancer treatments available to patients and, in particular may give options and hope to patients who have already failed on one or more therapies.
  • a combination cancer therapy comprising a HER inhibitor and a DHODH inhibitor.
  • a combination cancer therapy comprising a HER inhibitor and a DHODH inhibitor.
  • therapy comprises administering a therapeutic effect amount of a HER inhibitor and a DHODH inhibitor.
  • HER inhibitor and DHODH inhibitor for use in the manufacture of a combination therapy for the treatment of cancer, in particular a cancer described herein.
  • a HER2 inhibitor including a pan-HER inhibitor, such as Varlitinib co-administered with a DHODH inhibitor.
  • Varlitinib is administered in a 28 day cycle.
  • the therapy continues for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60 months or more.
  • DHODH is a key enzyme in the production of uridine, which is a central building block in the cell. Whilst not wishing to be bound by theory it may be that the DHODH inhibitor is able to upregulate p53 based apoptosis.
  • the up-regulation of p53 (which may lead to cell cycle arrest and at higher levels of p 53, to apoptosis) is likely to occur via mechanisms sensing the levels of intracellular uridine, and then setting forth a series of reactions leading to stabilisation of p53, and increasing its concentration.
  • a HER inhibitor such as a HER2 inhibitor or pan-HER inhibitor in the combination therapy of the present disclosure may be particular beneficial to minimises the cancers ability to resist treatment, in particular by "attacking" the cancer cells by two or more mechanisms with the combination therapy.
  • a method of treating a patient comprising administering a therapeutically effective amount of an inhibitor of at least HER2 and a therapeutically effective amount of DHODH inhibitor.
  • the HER2 inhibitor is a pan-HER inhibitor.
  • the HER2 inhibitor or pan-HER inhibitors is an organic chemistry molecule, for example with a molecular weight of 500 or less.
  • pan-HER inhibitor has a molecular formula of formula (I)
  • A is bonded to at least one of the carbons at positions 5, 6, 7 or 8 of the bicyclic ring and wherein the bicyclic ring is substituted by zero, one or two independent R 3 groups;
  • X is N, CH, CF or C-CN
  • A is Q or Z
  • R 2 is H or a substituted or unsubstituted Ci-e alkyl, allyl, substituted benzyl;
  • R 3 is hydrogen, halogen, cyano, nitro, Ci-Cio alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C3-C10 cycloalkyl, C3-C10 cycloalkylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, partially unsaturated heterocylyl, heterocyclylalkyl, -NR 4 S0 2 R 5 -S0 2 NR 6 R 4 , -C(0) R 6 , -C(0)OR 6 , -OC(0) R 6 , -NR 4 C(0)OR 5 , -NR 4 C(0)R 6 , -C(0)NR 4 R 6 , -NR 4 R 6 , -NR 4 C(0)NR 4 R 6 , -OR 6 , -S(0)R 5 , -SO2R 5 , or SR 6 , wherein said alkyl, alkenyl, alkyn
  • R 10 is hydrogen, halogen, cyano, nitro, C 1-C 10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C3-C10 cycloalkyl, C3-C10 cycloalkylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, partially unsaturated heterocyclyl, -NR 4 S0 2 R 5 , -S0 2 NR 6 R 4 , -C(0) R 6 , -C(0)OR 6 -OC(0)R 6 , -NR 4 C(0)OR 5 , -NR 4 C(0) R 6 , -C(0)NR 4 R 6 , -NR 4 R 6 , -NR 4 C(0)NR 4 R 6 , -OR 6 , -S(0) R 5 , -SO2R 5 , or SR 6 , wherein said alkyl, alkenyl, alkynyl
  • R 10 groups may be independently joined together with the atoms to which they are attached to complete a 3 to 10 membered cycloalkyl ring or heterocycloalkyl ring optionally containing one or more additional heteroatoms selected from the group consisting of 0, S, SO, SO2 and NR 6 , where each ring carbon is optionally substituted with one to three groups independently selected from the group consisting of halogen, C1-C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C3-C10 cycloalkyl, C3-C10 cycloalkylalkyl, cyano, nitro, trifluoromethyl, difluoromethoxy, trifluoromethoxy, azido, aryl, OR 8 , NR 6 R 8 , SR 6 , heteroaryl, arylalkyl, heteroarylallcyl, heterocyclyl, and heterocyclylalkyl, provided said ring does not contain
  • Z further includes:
  • Z includes one or more R 8 or R 9 groups, wherein said R 8 and R 9 groups may be bonded to the same or different atoms;
  • W and V are independently selected from the group consisting of CR 7 R 8 , CR 8 R 9 , 0, NR 6 , S, SO, and S0 2 ;
  • Y is selected from the group consisting of S, SO, SO 2 , CR 7 CR 8 , and CR 8 R 9 , optionally provided that when W is 0, NR 6 , S, SO, or S0 2 , then V is CR 8 R 9 , and
  • R 4 is H or Ci-6 alkyl
  • R 5 is trifluoromethyl, C 1 -C 1 0 alkyl, C3-C 1 0 cycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, or partially unsaturated heterocycle, wherein said alkyl, cycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, partially unsaturated heterocyclyl, and heterocyclylalkyl is optionally substituted with one to five groups independently selected from the group consisting of oxo, halogen, C 1 -C 1 0 alkyl, C 2 -C 1 0 alkenyl, C 2 -C 1 0 alkynyl, C3-C 1 0 cycloalkyl, C3-C 1 0 cycloalkylallcyl, cyano, nitro, OR 6 , NR 4 R 6 , SR
  • R 6 , R 8 and R 9 are independently selected from the group consisting of hydrogen, trifluoromethyl, C1-C10 alkyl, (CH2)o-4C3-Cio cycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, partially unsaturated heterocyclyl and heterocyclylalkyl, wherein said alkyl, cycloalkyl, aryl, arylalkyl, heteroaryl, heterocyclyl, partially unsaturated heterocyclyl, and heterocyclylalkyl is optionally substituted with one to five groups independently selected from the group consisting of oxo, halogen, C 1 -C 1 0 alkyl, C 2 -C 1 0 alkenyl, C 2 -C 1 0 alkynyl, C3-C 1 0 cycloalkyl, C3-Ciocycloalkylallcyl, cyano, nitro, OR 6
  • R 7 is hydrogen, halogen, cyano, nitro, C1-C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C3-C10 cycloalkyl, C3-C 1 0 cycloalkylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl, partially unsaturated heterocycle, -NR 4 S0 2 R 5 -S0 2 NR 6 R 4 , -C(0) R 6 , -C(0)OR 6 , -OC(0)R 6 , -NR 4 C(0)OR 5 , -NR 4 C(0)R 6 , -C(0)NR 4 R 6 , -NR 4 R 6 , -NR 4 C(0)NR 4 R 6 , -OR 6 , -S(0)R 5 , -SO2R 5 , or SR 6 , wherein said alkyl, alkenyl, alkyny
  • R 4 and R 6 together with the atoms to which they are attached may be independently joined to complete a 3 to 10 membered cycloalkyl ring or heterocycloalkyl ring optionally containing one or more additional heteroatoms selected from the group consisting of 0, S, SO, S0 2 and NR 6 , wherein each ring carbon is optionally substituted with one to three groups independently selected from the group consisting of halogen, C 1-C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C3-C10 cycloalkyl, C3-C 10 cycloalkylalkyl, cyano, nitro, trifluoromethyl, difluoromethoxy, trifluoromethoxy, azido, aryl, OR 8 , NR 6 R 8 , SR 6 , heteroaryl, arylalkyl, heteroarylalkyl, heterocyclyl, partially unsaturated heterocyclyl and heterocyclylalkyl, provided said
  • R 7 and R 8 together with the atoms to which they are attached may be independently joined to complete a 3 to 10 membered cycloalkyl ring or heterocycloalkyl ring optionally containing one or more additional heteroatoms selected from the group consisting of O, S, SO, SO2 and NR 6 , wherein each ring carbon is optionally substituted with one to three groups independently selected from the group consisting of halogen, C 1-C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C3-C10 cycloalkyl, C3-C 10 cycloalkylalkyl, cyano, nitro, trifluoromethyl, difluoromethoxy, trifluoromethoxy, azido, aryl, OR 8 , NR 6 R 8 , SR 6 , heteroaryl, arylalkyl, heteroarylalkyl, heterocyclyl, partially unsaturated heterocyclyl and heterocyclylalkyl; provided said
  • R 6 and R 10 together with the atoms to which they are attached may be independently joined to complete a 3 to 10 membered cycloalkyl ring or heterocycloalkyl ring optionally containing one or more additional heteroatoms selected from the group consisting of 0, S, SO, SO2 and NR 6 , wherein each ring carbon maybe optionally substituted with one to three groups independently selected from the group consisting of halogen, C1-C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C3-C10 cycloalkyl, C3-C10 cycloalkylalkyl, cyano, nitro, trifluoromethyl, difluoromethoxy, trifluoromethoxy, azido, aryl, OR 8 , NR 6 R 8 , SR 6 , heteroaryl, arylalkyl, heteroarylalkyl, heterocyclyl, partially unsaturated heterocyclyl and heterocyclylalkyl, provided said
  • R 8 and R 10 together with the atoms to which they are attached may be independently joined to complete a 3 to 10 membered cycloalkyl ring or heterocycloalkyl ring optionally containing one or more additional heteroatoms selected from the group consisting of O, S, SO, SO2 andNR 6 , wherein each ring carbon maybe optionally substituted with one to three groups independently selected from halogen, C1-C10 alkyl, C2-C10 alkenyl, C2-C 10 alkynyl, C3-C10 cycloalkyl, C3-C10 cycloalkylalkyl, cyano, nitro, trifluoromethyl, difluoromethoxy, trifluoromethoxy, azido, aryl, OR 8 , NR 6 R 8 , SR 6 , heteroaryl, arylalkyl, heteroarylalkyl, heterocyclyl, partially unsaturated heterocyclyl and heterocyclylalkyl, provided said ring does not contain
  • the HER inhibitor is an inhibitor of HER1, HER2, HER3, HER4 or a combination of 2 or more of the same.
  • the HER2 inhibitor is a biological therapeutic agent, for example herceptin or pertuzumab or a combination thereof.
  • the HER2 inhibitor is an antibody drug-conjugate, for example ado- trastuzumab-emtansine.
  • the biological therapeutic agent is administered parenterally.
  • pan-HER inhibitor is a compound of formula (la):
  • pan-HER inhibitor is Varlitinib: or a pharmaceutically acceptable salt thereof.
  • Varlitinib is employed as a free base.
  • Varlitinib at an appropriate dose is capable of inhibiting HERl, HER2 and HER4 directly and thought to be capable of inhibiting HER3 indirectly.
  • the compound of formula (I) (including formula (la) and Varlitinib) at least inhibits the activity of HERl and HER2, HERl and HER4 or HER2 and HER4.
  • the compound of formula (I) (including formula (la) and Varlitinib) at least inhibits the activity of HERl, HER2 and HER4, for example directly inhibits the activity of HERl, HER2 and HER4.
  • the compound of formula (I) (including formula (la) and Varlitinib) inhibits the activity of HERl, HER2, HER3 and HER4, for example directly inhibits the activity of HERl, HER2, and HER4, and indirectly inhibits the activity of HER3
  • each dose of the compound of formula (I), (including formula (la) and Varlitinib) is in the range 100 to 900mg, for example each dose is in the range of 300 to 500mg, such as 400mg, for example administered once or twice daily, such as twice daily.
  • patients may benefit from having the initial dose reduced to 300mg or 200mg bi-daily.
  • Varlitinib for in a regime which is non-continuous, for example taking medication on alternate days instead of each day or taking medication for four sequential days followed by one, two or three days without medication.
  • the compound of formula (I), (including formula (la) and Varlitinib) is administered orally.
  • the HER inhibitor is a combination of HER inhibitors, for example a combination of Varlitinib and Herceptin (trastuzumab) and/or pertuzumab.
  • a combination of Varlitinib and Herceptin show more therapeutic activity than either entity alone.
  • the HER inhibitor is a combination of ado-trastuzuma-emtansine and Varlitinib.
  • the DHODH inhibitor is selected from the group comprising teriflunomide, leflunomide a compound of formula (II) (disclosed in WO2008/077639 incorporated herein by reference) :
  • one of the groups G 1 represents a nitrogen atom or a group CR C and the other group represents
  • G 2 represents a nitrogen atom or a group CR d ;
  • R 1 represents a group selected from hydrogen, halogen, C 1- alkyl which may be optionally substituted with 1, 2 or 3 substituents selected from the group comprising halogen, hydroxy, and C3-8 cycloalkyl which may be optionally substituted with 1, 2 or 3 substituents selected from halogen and hydroxyl;
  • R 2 represents a group selected from hydrogen, halogen, hydroxyl, C1-4 alkyl which may be optionally substituted by 1, 2 or 3 substituents selected from the group comprising halogen, hydroxy, C3-8 alkyl which may be optionally substituted with 1, 2, or 3 substituents selected from halogen and hydroxyl;
  • R a , R b and R c independently represent a radical selected from the group comprising hydrogen, halogen, C 1-4 alkyl optionally substituted by 1, 2 or 3 substituents selected from the group comprising halogen, hydroxy and C 1.4 alkoxy;
  • R d represents a group selected from hydrogen, halogen, hydroxyl, C1-4 alkyl which may be substituted by 1, 2 or 3 substituents selected from the group comprising halogen, hydroxyl, C1-4 alkoxy which may be optionally substituted with 1, 2 or 3 substituent selected from the group comprising halogen, hydroxy, and C3-8 cycloalkoxy which may be optionally substituted with 1, 2 or 3 substitutents selected from halogen and hydroxyl;
  • G 3 & G 4 one is a nitrogen atom and the other is a CH;
  • M is hydrogen or a pharmaceutically acceptable cation.
  • R d represents a group selected from C1-4 alkoxy which may be optionally substituted with 1, 2 or 3 substituents selected from halogen, hydroxy, C3-8 cycloalkoxy which may be optionally substituted with 1, 2 or 3 substituents selected from halogen and hydroxyl.
  • the DHODH inhibitor is 2-(3, 5-difluoro-3'-methoxybiphenyl-4- ylamino) nicotinic acid (referred to herein as ASLAN003) or a pharmaceutically acceptable salt thereof, in particular:
  • the DHODH inhibitor is administered daily, for example once daily. In one embodiment the DHODH inhibitor is administered orally.
  • the DHODH inhibitor and HER inhibitor are administered sequentially in a treatment regimen, for example are administered on the same day.
  • the HER inhibitor such as Varlitinib is administered twice daily, for example a dose in the range disclosed herein.
  • the DHODH inhibitor and the HER inhibitors are administered simultaneously, at approximately the same time.
  • the DHODH inhibitor is administered in regimen that is daily or weekly for a continuous period of time for example 1 to 60 months or more, and the HER2 inhibitor or pan-HER inhibitor is administered intermittently during this period, for example Varlitinib may be administered in a one or more 28 days cycles.
  • Herceptin is an antibody molecule, such as Herceptin
  • the administration protocol is likely to be very different to that of small molecule inhibitors.
  • Herceptin for example may be administered (in particular in combination with cytotoxic chemotherapy) in a regime as follows:
  • the HER2 inhibitor or pan-HER inhibitor is administered in regimen that is daily or weekly for a continuous period of time, for example 1 to 60 months or more, and the DHODH inhibitor is administered intermittently during this period.
  • Administered intermittently as employed herein refers to a period wherein the therapy is administered and then stopped with the option of starting the therapy again at some point in the future.
  • the DHODH inhibitor is administered in regimen that is daily or weekly for a continuous period of time, for example 1 to 60 months or more
  • the HER2 inhibitor or pan-HER inhibitor is administered in regimen that is daily or weekly for a continuous period of time, for example 1 to 60 months or more.
  • the DHODH inhibitor is administered in regimen that is daily or weekly for intermittent periods over for example 1 to 60 months or more, and the HER2 inhibitor or pan-HER inhibitor is administered conjunctly with DHODH inhibitor in regimen that is daily or weekly for intermittent periods over, for example 1 to 60 months or more.
  • the DHODH inhibitor and the HER2 or pan-HER inhibitor are co- formulated.
  • the DHODH inhibitor is administered orally.
  • the HER inhibitor is administered orally, parenterally or both, in particular orally.
  • the HER inhibitor such as HER2 inhibitor is administered orally or parenterally, for example intravenously.
  • the HER inhibitor such as pan-HER inhibitor is administered orally.
  • one is administered orally and one is administered parenterally, such as intravenously.
  • the DHODH inhibitor and the pan-HER inhibitor are both administered orally.
  • the cancer is selected from liver cancer, biliary tract cancer, breast cancer (such as none ER+ breast cancer), prostate cancer, colorectal cancer, ovarian cancer, cervical cancer, lung cancer, gastric cancer, pancreatic, bone cancer, bladder cancer, head and neck cancer, thyroid cancer, skin cancer, renal cancer, and oesophagus cancer, for example gastric cancer, hepatocellular carcinoma and cholangiocarcinoma.
  • the cancer is selected from and epithelial cancer, for example selected from the group comprising hepatocellular carcinoma, cholangiocarcinoma, breast cancer, prostate cancer, colorectal cancer, ovarian cancer, lung cancer, stomach cancer, pancreatic and oesophagus cancer.
  • the cancer is selected from other cancer forms where HER inhibition is effective.
  • the patient population for treatment has HER pathway activation indicated by high levels of phosphorylated downstream signalling proteins, for example selected from pAKT and pERK.
  • the patient is a human.
  • the combination therapy of the present disclosure is efficacious and, for example beneficial in that it provides augmented therapeutic activity in comparison to monotherapy comprising one of the components.
  • Augmented activity may be any beneficial therapeutic effect of employing the combination of the present disclosure, for example an increase in anti-tumor activity and/or a reduced propensity for the cancer to become resistant.
  • Other benefits may be therapeutic effect in patients who have failed one or more lines of therapy.
  • the patient population has a cancer that is resistant or refractory to known therapies, such as cytotoxic chemotherapy.
  • Combination therapy refers to two or more modes of therapy being employing over the same treatment period, i.e. the opposite of sequential therapy.
  • Two or more modes of therapy as employed herein refers to at least two therapies which have different modes of action and/or different activities and/or different routes of administration.
  • the HER inhibitor and the DHODH inhibitor has to administered in a time frame, where the pharmacological effects of a HER inhibitor and the DHODH inhibitor overlap, i.e. the treatment regimens for the said therapies partly coincide in time.
  • the treatment regimens for the said therapies partly coincide in time.
  • Combination therapy refers to where a medicament according to the present disclosure is administered in a treatment regimen along with at least one further therapeutic agent.
  • the regime may be separate formulations administered at the same time or different times or co-formulations of the two or more therapeutic agents.
  • the "first" medicament employed in the combination therapy according to the present disclosure may be administered; prior to the further therapeutic agent or agents, concomitant with the further therapeutic agent or agents, or after the further therapeutic agent or agents.
  • therapeutic agent or agents such as an anti-cancer therapy are employed in combination with the therapy of the present disclosure.
  • the therapeutic agent is a chemotherapeutic agent.
  • Chemotherapeutic agent as employed herein is intended to refer to specific antineoplastic chemical agents or drugs that are destructive to malignant cells and tissues, including alkylating agents, antimetabolites, anthracyclines, plant alkaloids, topoisomerase inhibitors, and other antitumour agents.
  • Specific examples of chemotherapy include doxorubicin, 5-fluorouracil (5-FU), paclitaxel (for example abraxane or docetaxel), capecitabine, irinotecan, and platins, such as cisplatin and oxaliplatin or a combination thereof.
  • a suitable dose may be chosen by the practitioner based on the nature of the cancer being treated and the patient.
  • Co-administered as employed herein refers to administration of the HER inhibitor and DHODH inhibitor at the same time or approximately the same time (including where the actives are administered by the same or different routes).
  • Inhibitor refers to the reduction of a relevant biological activity, for example by 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 or 100%, such as when measured in a relevant in vitro assay.
  • Direct inhibition is where the inhibitor binds directly to or physically blocks a binding interaction to inhibit a biological activity, or when the inhibitor inhibits the activation through phosphorylation of the target molecule.
  • Indirect inhibition refers to where the biological activity in question is inhibited as a result of directly inhibiting a target that is other than the entity that is indirectly inhibited.
  • Dihydroorotate dehydrogenase is the enzyme that catalyzes the fourth step in the pyrimidine biosynthetic pathway namely the conversion of dihydroorotate to orotate concomitantly with an electron transfer to ubiquinone (cofactor Q) via a flavin mononucleotide intermediate (Loffler Mol Cell Biochem, 1997).
  • DHODH Dihydroorotate dehydrogenase
  • mitochondrial cytochrome bcl a component of the electron transport chain complex III
  • DHODH dihydroorotate dehydrogenase
  • a DHODH as employed herein refers to a compound which inhibits the activity of dihydroorotate dehydrogenase, in particular in vivo.
  • Compounds of formula (II) described above are examples of DHODH inhibitors. These compounds are disclosed in WO2008/077639, incorporated herein by reference.
  • DHODH inhibitor which may be employed in a methods of the present disclosure include:
  • Suitable salts of DHODH inhibitors include those disclosed in WO2010/102826, WO2010/10225 and WO2010/102824 each incorporated herein by reference.
  • An inhibitor of at least HER2 refers to therapeutic agent, for example biological therapy or a "drug", which inhibits HER2 but may also inhibit other HER receptors, for example the group comprising HER1, HER3, HER4 and combinations thereof.
  • Pan-HER inhibitor as employed herein refers to a molecule that inhibits at least two molecules from the ErbB family of proteins, namely ErbB-1 (also known as HER1 and EGFR), ErbB-2 (HER2), ErbB-3 (HER3), and ErbB-4(HER4).
  • pan-HER inhibitor as employed herein refers to a therapeutic agent, for example a chemical entity, which inhibits at least two HER receptors, for example an inhibitor of HER 1 and HER2.
  • a biological therapeutic is one based on a protein, for example an antibody or binding fragment thereof, including fusion proteins and biological molecules conjugated to a polymer, toxin or similar payload.
  • a “drug” as employed herein refers to a chemical entity, organic chemistry molecule with pharmacological activity.
  • the HER2 inhibitor is a specific HER2 inhibitor, for example a monoclonal antibody specific to HER2 and described in W092/22653 incorporated herein by reference, such as herceptin.
  • An example of a biological therapeutic conjugated to a payload, suitable for use in the therapy of the present disclosure, is trastuzumab emtansine.
  • the HER2 inhibitor is a HER dimerization inhibitor, for example pertuzumab disclosed in WO01/00244 and WO01/100245 incorporated herein by reference.
  • the pan-HER inhibitor is a compound of formula (I) or (la) described above and disclosed in WO2005/016346 incorporated herein by reference, in particular (fi)-N4- [3-Chloro-4-(thiazol-2-ylmethoxy)-phenyl]-N6-(4-methyl-4,5,-dihydro-oxazol-2-yl)-quinazoline- 4,6-diamine (Varlitinib) or a pharmaceutically acceptable salt thereof.
  • Varlitinib is employed as free base.
  • the treatment of the present disclosure is administered for epithelial cancer, for example is selected from liver cancer (such as hepatocellular carcinoma), biliary tract cancer, breast cancer (such as none ER+ breast cancer), prostate cancer, colorectal cancer, ovarian cancer, cervical cancer, lung cancer, gastric cancer, pancreatic, bone cancer, bladder cancer, head and neck cancer, thyroid cancer, skin cancer, renal cancer, and oesophagus cancer, for example gastric cancer.
  • liver cancer such as hepatocellular carcinoma
  • breast cancer such as none ER+ breast cancer
  • prostate cancer colorectal cancer
  • ovarian cancer cervical cancer
  • lung cancer gastric cancer
  • pancreatic bone cancer
  • bladder cancer head and neck cancer
  • thyroid cancer skin cancer
  • renal cancer renal cancer
  • oesophagus cancer for example gastric cancer.
  • the cancer is selected from selected from the group comprising hepatocellular carcinoma, cholangiocarcinoma, breast cancer, prostate cancer, colorectal cancer, ovarian cancer, lung cancer, gastric cancer, pancreatic and oesophagus cancer.
  • the biliary duct cancer is in a location selected from intrahepatic bile ducts, left hepatic duct, right hepatic duct, common hepatic duct, cystic duct, common bile duct, Ampulla of Vater and combinations thereof.
  • the biliary duct cancer is in an intrahepatic bile duct.
  • the biliary duct cancer is in a left hepatic duct.
  • the biliary duct cancer is in a right hepatic duct.
  • the biliary duct cancer is in a common hepatic duct.
  • the biliary duct cancer is in a cystic duct.
  • the biliary duct cancer is in a common bile duct.
  • the biliary duct cancer is in an Ampulla of Vater.
  • the epithelial cancer is a carcinoma.
  • the combination treatment according to the disclosure is adjuvant therapy, for example after surgery.
  • the combination therapy according to the disclosure is neoadjuvant treatment, for example to shrink a tumour before surgery.
  • the tumour is a solid tumour.
  • the cancer is a primary cancer, secondary cancer, metastasis or combination thereof.
  • the treatment according to the present disclosure is suitable for the treatment of secondary tumours.
  • the cancer is metastatic cancer.
  • the treatment according to the present disclosure is suitable for the treatment of primary cancer and metastases. .
  • the treatment according to the present disclosure is suitable for the treatment of secondary cancer and metastases. .
  • the treatment according to the present disclosure is suitable for the treatment of primary cancer, secondary cancer and metastases.
  • the treatment according to the present disclosure is suitable for the treatment of cancerous cells in a lymph node, for a cancer of the present disclosure.
  • the liver cancer is primary liver cancer. In one embodiment the liver cancer is secondary liver cancer. In one embodiment the liver cancer is stage 1, 2, 3 A, 3B, 3C, 4A or 4B.
  • the gastric cancer is stage 0, 1, II, III or IV.
  • the cancer is EGFR (HERl) positive.
  • the patient population is HER2 positive or HER2 amplified.
  • Cancers where HER2 is known to be overexpressed include, but are not limited to, breast cancers, gastric cancer, esophageal cancer, ovarian cancer, endometrial cancer, bladder cancer, colorectal cancer, lung cancer, and head and neck cancers.
  • the patient is selected for HER2 expression or overexpression, for example before starting the combination therapy according to the present disclosure.
  • the cancer has unknown HER2 expression.
  • the patient population is EGFR and HER2 positive.
  • Examples of pharmaceutically acceptable salts include but are not limited to acid addition salts of strong mineral acids such as HC1 and HBr salts and addition salts of strong organic acids, such as a methansulfonic acid salt, tosylates, furoates and the like, including di, tri salts thereof, such as ditosylates.
  • the combination therapy according to the present disclosure further comprises a RON inhibitor, for example as disclosed WO2008/058229, incorporated herein by reference.
  • the combination therapy of the present disclosure comprises a checkpoint inhibitor, such as a CTLA4 inhibitor, a PD-1 inhibitor or a PD-L1 inhibitor, in particular an antibody or binding fragment thereof.
  • a checkpoint inhibitor such as a CTLA4 inhibitor, a PD-1 inhibitor or a PD-L1 inhibitor, in particular an antibody or binding fragment thereof.
  • the combination therapy of the present disclosure further comprises a chemotherapeutic agent.
  • the gastric cancer is selected from the group comprising adenocarcinoma of the stomach, squamous cell carcinomas, lymphoma of the stomach, gastric stromal tumor, and neuroendocrine tumors.
  • the liver cancer is, for example selected from the group hepatocellular carcinoma, cholangiocarcinoma, angiosarcoma, and hepatoblastoma, in particular hepatocellular carcinoma.
  • the primary liver cancer is stage 1, 2, 3 or 4.
  • the liver cancer is secondary or metastasized liver cancer.
  • Prostate cancer refers to cancer of the prostate, for example ductal adenocarcinoma, transitional cell (urothelial cancer), squamous cell cancer, carcinoid of the prostate, small cell cancer or sarcoma and sarcomatoid cancer.
  • Pancreatic cancer as employed herein includes exocrine cancers (including rare forms thereof such as cystitic tumours, and cancer of the acinar cells), endocrine pancreatic tumours (including gastrinomas, insulinomas, somatostatinomas, VIPomas, glucagonomas), pancreatoblastoma, sarcomas of the pancreas and lymphoma.
  • Biliary tract cancer refers to cholangiocarcinoma (intrahepatic, extrahepatic), gall bladder cancer and ampullary carcinoma.
  • Colorectal cancer refers to cancer or the colon and/or rectum and includes squamous cell cancers, carcinoid tumours, sarcomas and lymphomas.
  • Breast cancer refers to cancer of the breast and includes ductal cardinoma in situ, lobular carcinoma in situ, invasive ductal breast cancer, invasive lobular breast cancer, invasive breast cancer, Paget's disease, angiosarcoma of the breast and rare types of breast cancer such as medullary breast cancer, mucinous breast cancer, tubular breast cancer, adenoid cystic carcinoma of the breast metaplastic breast cancer, basal type breast cancer and papillary breast cancer.
  • Lung cancers are classified according to histological type and are categorized by the size and appearance of the malignant cells seen by a histopathologist under a microscope. For therapeutic purpose, two broad classes are distinguished: non-small cell lung carcinoma and small cell lung carcinoma.
  • the epithelial cancer is lung cancer, for example small-cell lung cancer (SCLC) and non-small-cell lung cancer (NSCLC).
  • Non-small-cell lung carcinoma The three main subtypes of NSCLC are adenocarcinoma, squamous-cell carcinoma and large-cell carcinoma.
  • adenocarcinoma Nearly 40% of lung cancers are adenocarcinoma, which usually originates in peripheral lung tissue.
  • a subtype of adenocarcinoma, the bronchioloalveolar carcinoma, is more common in female never-smokers, and may have a better long term survival.
  • Squamous-cell carcinoma accounts for about 30% of lung cancers. They typically occur close to large airways. A hollow cavity and associated cell death are commonly found at the center of the tumor. About 9% of lung cancers are large-cell carcinoma. These are so named because the cancer cells are large, with excess cytoplasm, large nuclei and conspicuous nucleoli.
  • SCLC Small-cell lung carcinoma-In small-cell lung carcinoma
  • the cells contain dense neurosecretory granules (vesicles containing neuroendocrine hormones), which give this tumor an endocrine/paraneoplastic syndrome association.
  • These cancers grow quickly and spread early in the course of the disease. Sixty to seventy percent have metastatic disease at presentation.
  • the cancer is non-small lung carcinoma.
  • the cancer is liver cancer, for example a liver metastasis from a primary cancer, for example colon cancer, which has spread to the liver.
  • the liver cancer is HCC hepatocellular carcinoma.
  • renal cancer for example renal cell carcinoma and/or urothelial cell carcinoma.
  • Other examples of renal cancer include squamous cell carcinoma, juxtaglomerular cell tumor (reninoma), angiomyolipoma, renal oncocytoma,
  • Bellini duct carcinoma clear-cell sarcoma of the kidney, mesoblastic nephroma, Wilms' tumor, mixed epithelial stromal tumor, clear cell adenocarcinoma, transitional cell carcinoma, inverted papilloma, renal lymphoma, teratoma, carcinosarcoma, and carcinoid tumor of the renal pelvis.
  • the cancer is bladder cancer, for example is any of several types of malignancy arising from the epithelial lining (i.e., the urothelium) of the urinary bladder.
  • the epithelial lining i.e., the urothelium
  • bladder cancers are transitional cell carcinoma.
  • the other 10% are squamous cell carcinoma, adenocarcinoma, sarcoma, small cell carcinoma, and secondary deposits from cancers elsewhere in the body.
  • the staging of is given below.
  • the current disclosure extends to any stage of bladder cancer.
  • Cancerous ovarian tumors can start from three common cell types:
  • the present disclosure relates to treatment of ovarian cancer from any source, for example as described herein, in particular epithelium cells.
  • Epithelial ovarian carcinomas account for 85 to 90 percent of all cancers of the ovaries.
  • Epithelial ovarian tumors develop from the cells that cover the outer surface of the ovary. Most epithelial ovarian tumors are benign (noncancerous). There are several types of benign epithelial tumors, including serous adenomas, mucinous adenomas, and Brenner tumors. Cancerous epithelial tumors are carcinomas - meaning they begin in the tissue that lines the ovaries. These are the most common and most dangerous of all types of ovarian cancers. Unfortunately, almost 70 percent of women with the common epithelial ovarian cancer are not diagnosed until the disease is advanced in stage.
  • LMP tumors tumors of low malignant potential
  • Germ Cell Tumors - Ovarian germ cell tumors develop from the cells that produce the ova or eggs. Most germ cell tumors are benign (non-cancerous), although some are cancerous and may be life threatening. The most common germ cell malignancies are maturing teratomas, dysgerminomas, and endodermal sinus tumors. Germ cell malignancies occur most often in teenagers and women in their twenties. Today, 90 percent of patients with ovarian germ cell malignancies can be cured and their fertility preserved.
  • Stromal Tumors - Ovarian stromal tumors are a rare class of tumors that develop from connective tissue cells that hold the ovary together and those that produce the female hormones, estrogen and progesterone.
  • the most common types are granulosa-theca tumors and Sertoli- Leydig cell tumors. These tumors are quite rare and are usually considered low-grade cancers, with approximately 70 percent presenting as Stage I disease (cancer is limited to one or both ovaries).
  • Primary Peritoneal Carcinoma The removal of one's ovaries eliminates the risk for ovarian cancer, but not the risk for a less common cancer called Primary Peritoneal Carcinoma.
  • Peritoneal Carcinoma is closely rated to epithelial ovarian cancer (most common type). It develops in cells from the peritoneum (abdominal lining) and looks the same under a microscope. It is similar in symptoms, spread and treatment.
  • stage of a tumor can be determined during surgery, when the doctor can tell if the cancer has spread outside the ovaries.
  • the treatment plan and prognosis (the probable course and outcome of your disease) will be determined by the stage of cancer you have.
  • Stage I - Growth of the cancer is limited to the ovary or ovaries.
  • Stage IA - Growth is limited to one ovary and the tumor is confined to the inside of the ovary.
  • Stage IB - Growth is limited to both ovaries without any tumor on their outer surfaces. There are no ascites present containing malignant cells. The capsule is intact.
  • Stage IC The tumor is classified as either Stage IA or IB and one or more of the following are present: (1) tumor is present on the outer surface of one or both ovaries; (2) the capsule has ruptured; and (3) there are ascites containing malignant cells or with positive peritoneal washings.
  • Stage II - Growth of the cancer involves one or both ovaries with pelvic extension.
  • Stage IIA The cancer has extended to and/or involves the uterus or the fallopian tubes, or both.
  • Stage IIC The tumor is classified as either Stage IIA or IIB and one or more of the following are present: (1) tumor is present on the outer surface of one or both ovaries; (2) the capsule has ruptured; and (3) there are ascites containing malignant cells or with positive peritoneal washings.
  • Stage III - Growth of the cancer involves one or both ovaries, and one or both of the following are present: (1) the cancer has spread beyond the pelvis to the lining of the abdomen; and (2) the cancer has spread to lymph nodes. The tumor is limited to the true pelvis but with histologically proven malignant extension to the small bowel or omentum.
  • Stage IIIA - During the staging operation, the practitioner can see cancer involving one or both of the ovaries, but no cancer is grossly visible in the abdomen and it has not spread to lymph nodes. However, when biopsies are checked under a microscope, very small deposits of cancer are found in the abdominal peritoneal surfaces.
  • Stage IIIB The tumor is in one or both ovaries, and deposits of cancer are present in the abdomen that are large enough for the surgeon to see but not exceeding 2 cm in diameter. The cancer has not spread to the lymph nodes.
  • Stage IIIC The tumor is in one or both ovaries, and one or both of the following is present: (1) the cancer has spread to lymph nodes; and/or (2) the deposits of cancer exceed 2 cm in diameter and are found in the abdomen.
  • Stage IV This is the most advanced stage of ovarian cancer. Growth of the cancer involves one or both ovaries and distant metastases (spread of the cancer to organs located outside of the peritoneal cavity) have occurred. Finding ovarian cancer cells in pleural fluid (from the cavity which surrounds the lungs) is also evidence of stage IV disease.
  • the ovarian cancer is: type I, for example IA, IB or IC; type II, for example IIA, IIB or IIC; type III, for example IIIA, IIIB or IIIC; or type IV.
  • the breast cancer is one selected from the group comprising ductal carcinoma in situ, lobular carcinoma in situ, invasive breast cancer, invasive lobular breast cancer, Paget's disease, angiosarcoma of the breast, medulllary breast cancer, mucinous breast cancer, tubular breast cancer, adenoid cystic carcinoma of the breast, metaplastic breast cancer, lymphoma of the breast, basal type breast cancer, phyllodes or cystosarcoma phyllodes and papillary breast cancer.
  • the prostate cancer is selected from the group comprising ductal adenocarcinoma, transitional cell (urothelial) cancer, squamous cell cancer, carcinoid, small cell cancer, sarcomas and sarcomatoid cancers.
  • Thyroid cancer refers to cancer of the thyroid originating from follicular or parafollicular thyroid cells and includes papillary thyroid cancer (75% to 85% of cases); follicular thyroid cancer (10% to 20% of cases); medullary thyroid cancer (5% to 8% of cases)- cancer of the parafollicular cells, often part of multiple endocrine neoplasia type 2; poorly differentiated thyroid cancer; anaplastic thyroid cancer (less than 5% of cases) is not responsive to treatment and can cause pressure symptoms, thyroid lymphoma, squamous cell thyroid carcinoma, sarcoma of thyroid.
  • Renal cancer refers to cancer of the kidney, for example renal cell carcinoma and transitional cell carcinoma of the renal pelvis, such as squamous cell carcinoma, juxtaglomerular cell tumor (reninoma), angiomyolipoma, renal oncocytoma, bellini duct carcinoma, clear-cell sarcoma of the kidney, mesoblastic nephroma, Wilms' tumor, mixed epithelial stromal tumor, clear cell adenocarcinoma, transitional cell carcinoma, inverted papilloma, renal lymphoma, teratoma, carcinosarcoma; carcinoid tumor of the renal pelvis.
  • reninoma juxtaglomerular cell tumor
  • angiomyolipoma angiomyolipoma
  • renal oncocytoma bellini duct carcinoma
  • clear-cell sarcoma of the kidney mesoblastic nephroma, Wilms' tumor, mixed epithelial stromal tumor
  • Bladder cancer as employed herein refers to cancer of the bladder including transitional cell bladder cancer, carcinoma in situ, papillary cancer and rarer types of bladder cancer such as squamous cell cancer and adenocarcinoma.
  • Esophageal cancer refers to cancer of the oesphagus including esophageal squamous-cell carcinomas, esophageal adenocarcinomas, and variants of squamous- cell carcinoma, and non-epithelial tumors, such as leiomyosarcoma, malignant melanoma, rhabdomyosarcoma, lymphoma, among others.
  • Head and neck cancer refers to cancer of the neck and/or head, including mouth cancer, nasopharyngeal cancer, oropharyngeal cancer, paranasal sinus cancer and salivary gland cancer.
  • the combination therapy of the present disclosure may be employed in combination with a further cancer therapy, for example chemotherapy.
  • Chemotherapeutic agent and chemotherapy or cytotoxic agent are employed interchangeably herein unless the context indicates otherwise.
  • Chemotherapy as employed herein is intended to refer to specific antineoplastic chemical agents or drugs that are "selectively" destructive to malignant cells and tissues, for example alkylating agents, antimetabolites including thymidylate synthase inhibitors, anthracyclines, anti- microtubule agents including plant alkaloids, topoisomerase inhibitors, parp inhibitors and other antitumour agents. Selectively in this context is used loosely because of course many of these agents have serious side effects.
  • the preferred dose may be chosen by the practitioner, based on the nature of the cancer being treated.
  • alkylating agents which may be employed in the method of the present disclosure include an alkylating agent nitrogen mustards, nitrosoureas, tetrazines, aziridines, platins and derivatives, and non-classical alkylating agents.
  • Example a platinum containing chemotherapeutic agent (also referred to as platins), such as cisplatin, carboplatin, oxaliplatin, satraplatin, picoplatin, nedaplatin, triplatin and lipoplatin (a liposomal version of cisplatin), in particular cisplatin, carboplatin and oxaliplatin.
  • chemotherapeutic agent also referred to as platins
  • platins such as cisplatin, carboplatin, oxaliplatin, satraplatin, picoplatin, nedaplatin, triplatin and lipoplatin (a liposomal version of cisplatin), in particular cisplatin, carboplatin and oxaliplatin.
  • the dose for cisplatin ranges from about 20 to about 270 mg/m 2 depending on the exact cancer. Often the dose is in the range about 70 to about 100mg/m 2 .
  • Nitrogen mustards include mechlorethamine, cyclophosphamide, melphalan, chlorambucil, ifosfamide and busulfan.
  • Nitrosoureas include N-Nitroso-N-methylurea (MNU), carmustine (BCNU), lomustine
  • CCNU CCNU
  • MeCCNU semustine
  • Tetrazines include dacarbazine, mitozolomide and temozolomide.
  • Aziridines include thiotepa, mytomycin and diaziquone (AZQ).
  • antimetabolites examples include anti-folates (for example methotrexate and pemetrexed), purine analogues (for example thiopurines, such as azathiopurine, mercaptopurine, thiopurine, fludarabine (including the phosphate form), pentostatin and cladribine), pyrimidine analogues (for example fluoropyrimidines, such as 5-fluorouracil and prodrugs thereof such as capecitabine [Xeloda®]), floxuridine, gemcitabine, cytarabine, decitabine, raltitrexed(tomudex) hydrochloride, cladribine and 6-azauracil.
  • anti-folates for example methotrexate and pemetrexed
  • purine analogues for example thiopurines, such as azathiopurine, mercaptopurine, thiopurine, fludarabine (including the phosphate form),
  • anthracyclines examples include daunorubicin (Daunomycin), daunorubicin (liposomal), doxorubicin (Adriamycin), doxorubicin (liposomal), epirubicin, idarubicin, valrubicin currenlty used only to treat bladder cancer and mitoxantrone an anthracycline analog, in particular doxorubicin.
  • anti-microtubule agents examples include vinca alkaloids and taxanes.
  • Vinca alkaloids include completely natural chemicals for example vincristine and vinblastine and also semi-synthetic vinca alkaloids, for example vinorelbine, vindesine, and vinflunine
  • Taxanes include paclitaxel, docetaxel, abraxane, carbazitaxel and derivatives of thereof.
  • Derivatives of taxanes as employed herein includes reformulations of taxanes like taxol, for example in a micelluar formulaitons, derivatives also include chemical derivatives wherein synthetic chemistry is employed to modify a starting material which is a taxane.
  • Topoisomerase inhibitors which may be employed in a method of the present disclosure include type I topoisomerase inhibitors, type II topoisomerase inhibitors and type II topoisomerase poisons.
  • Type I inhibitors include topotecan, irinotecan, indotecan and indimitecan.
  • Type II inhibitors i ich has the following structure:
  • Type II poisons include amsacrine, etoposide, etoposide phosphate, teniposide and doxorubicin and fluoroquinolones.
  • the chemotherapeutic is a PARP inhibitor.
  • chemotherapeutic agents employed is, for example a platin and 5-FU or a prodrug thereof, for example cisplatin or oxaplatin and capecitabine or gemcitabine, such as FOLFOX.
  • the chemotherapy comprises a combination of chemotherapy agents, in particular cytotoxic chemotherapeutic agents.
  • the chemotherapy combination comprises a platin, such as cisplatin and fluorouracil or capecitabine.
  • the chemotherapy combination in capecitabine and oxaliplatin in capecitabine and oxaliplatin (Xelox).
  • the chemotherapy is a combination of folinic acid and 5-FU, optionally in combination with oxaliplatin.
  • the chemotherapy is a combination of folinic acid, 5-FU and irinotecan (FOLFIRI), optionally in combination with oxaliplatin (FOLFIRINOX).
  • the regimen consists of: irinotecan (180 mg/m 2 IV over 90 minutes) concurrently with folinic acid (400 mg/m 2 [or 2 x 250 mg/m 2 ] IV over 120 minutes); followed by fluorouracil (400-500 mg/m 2 IV bolus) then fluorouracil (2400-3000 mg/m 2 intravenous infusion over 46 hours). This cycle is typically repeated every two weeks.
  • the dosages shown above may vary from cycle to cycle.
  • the chemotherapy combination employs a microtubule inhibitor, for example vincristine sulphate, epothilone A, N-[2-[(4-Hydroxyphenyl)amino]-3-pyridinyl]-4- methoxybenzenesulfonamide (ABT-751), a taxol derived chemotherapeutic agent, for example paclitaxel, abraxane, or docetaxel or a combination thereof.
  • a microtubule inhibitor for example vincristine sulphate, epothilone A, N-[2-[(4-Hydroxyphenyl)amino]-3-pyridinyl]-4- methoxybenzenesulfonamide (ABT-751), a taxol derived chemotherapeutic agent, for example paclitaxel, abraxane, or docetaxel or a combination thereof.
  • the chemotherapy combination employs an mTor inhibitor.
  • mTor inhibitors include: everolimus (RAD001), WYE-354, KU-0063794, papamycin (Sirolimus), Temsirolimus, Deforolimus(MK-8669), AZD8055 and BEZ235(NVP-BEZ235).
  • the chemotherapy combination employs a MEK inhibitor.
  • MEK inhibitors include: AS703026, CI-1040 (PD184352), AZD6244 (Selumetinib), PD318088, PD0325901, AZD8330, PD98059, U0126-EtOH, BIX 02189 or BIX 02188.
  • the chemotherapy combination employs an AKT inhibitor.
  • AKT inhibitors include: MK-2206 and AT7867.
  • the combination employs an aurora kinase inhibitor.
  • aurora kinase inhibitors include: Aurora A Inhibitor I, VX-680, AZD1152-HQPA (Barasertib), SNS- 314 Mesylate, PHA-680632, ZM-447439, CCT129202 and Hesperadin.
  • the chemotherapy combination employs a p38 inhibitor, for example as disclosed in WO2010/038086, such as -[4-( ⁇ 4-[3-(3-ieri-Butyl-l-p-tolyl-lH-pyrazol-5- yl) ureido] naphthalen- 1 -yloxy ⁇ methyl) pyridin-2 -yl] - 2-methoxyacetamide.
  • a p38 inhibitor for example as disclosed in WO2010/038086, such as -[4-( ⁇ 4-[3-(3-ieri-Butyl-l-p-tolyl-lH-pyrazol-5- yl) ureido] naphthalen- 1 -yloxy ⁇ methyl) pyridin-2 -yl] - 2-methoxyacetamide.
  • the combination employs a Bcl-2 inhibitor.
  • Bcl-2 inhibitors include: obatoclax mesylate, ABT-737, ABT-263(navitoclax) and TW-37.
  • the chemotherapy combination comprises an antimetabolite such as capecitabine (xeloda), fludarabine phosphate, fludarabine (fludara), decitabine, raltitrexed (tomudex), gemcitabine hydrochloride and cladribine.
  • an antimetabolite such as capecitabine (xeloda), fludarabine phosphate, fludarabine (fludara), decitabine, raltitrexed (tomudex), gemcitabine hydrochloride and cladribine.
  • the chemotherapy combination comprises ganciclovir, which may assist in controlling immune responses and/or tumour vasculation.
  • the chemotherapy includes a PARP inhibitor.
  • one or more therapies employed in the method herein are metronomic, that is a continuous or frequent treatment with low doses of anticancer drugs, often given concomitant with other methods of therapy.
  • the combination of the present disclosure is employed after chemotherapy.
  • the combination therapy of the present disclosure is employed before chemotherapy.
  • the dose of chemotherapy employed in the combination therapy of the present disclosure is lower than the dose of chemotherapy employed in "monotherapy" (where monotherapy may include the dose of chemotherapy employed when combinations of chemotherapy agents are employed).
  • the medicament is administered in combination with therapy complimentary to the cancer therapy, for example a treatment for cachexia, such as cancer cachexia, for example S-pindolol, S-mepindolol or S-bopindolol.
  • a treatment for cachexia such as cancer cachexia, for example S-pindolol, S-mepindolol or S-bopindolol.
  • Suitable doses may be in the range of 2.5mg to lOOmg, such as 2.5mg to 50mg per day provided a single dose or multiple doses given as multiple doses administered during the day.
  • Treatment as employed herein refers to where the patient has a disease or disorder, for example cancer and the medicament according to the present disclosure is administered to stabilise the disease, delay the disease, amelorate the disease, send the disease into remission, maintain the disease in remission or cure the disease. Treating as employed herein includes administration of a medicament according to the present disclosure for treatment or prophylaxis.
  • the present disclosure is explained in the context of a method of treating a patients. However, the disclosure extends to use of the combination therapy as described herein for use in treatment, in particular for the treatment of cancer, such as a cancer described herein. Also provided is use of the combination of compounds as described herein for the manufacture of a medicament for the treatment of cancer, in particular a cancer described herein.
  • the combination therapy according to the present disclosure is employed as cancer adjuvant therapy, for example after surgery to remove some or all of the cancerous cells.
  • the combination according to the present disclosure is employed as neoadjuvant therapy, for example before surgery to remove some or all of the cancerous cells.
  • a therapeutically effective dose (such as a daily dose) of a DHODH inhibitor is in the range lOmg to lOOOmg, for example 50 to 500mg, such as 100, 150, 200, 250, 300, 350, 400, 450, 500mg, in particular administered once a day.
  • pan-HER inhibitor for example the compound of formula (I) or
  • (la) in particular Varlitinib is administered bi-daily, such as at a dose in the range lOOmg to 900mg on each occasion, in particular 300mg, or 400mg or 500mg each dose.
  • active ingredients employed in the therapy of the present disclosure will be provided in the form of a pharmaceutical formulation comprising one or more excipients, diluents or carriers.
  • the compound of formula (I), (la) or Varlitinib is administered as pharmaceutical formulation comprising one or more pharmaceutically acceptable excipients.
  • Embodiments of the invention comprising certain features/elements are also intended to extend to alternative embodiments "consisting" or “consisting essentially” of the relevant elements / features.
  • Varlitinib inhibits phosphorylation of the HER pathway and AKT signaling.
  • Varlitinib and ASLAN003 combo leads to reduction in HER3 protein levels
  • Varlitinib inhibits the Rb pathway and phosphorylation of cdc2.
  • pan-HER inhibitor 2-(3,5-difluoro-3'-methoxybiphenyl-4-ylamino)nicotinic acid (ASLAN003) and the pan-HER inhibitor is (fi)-N4-[3-Chloro-4-(thiazol-2-ylmethoxy)-phenyl]-N6-(4-methyl-4,5,-dihydro-oxazol- 2-yl)-quinazoline-4,6-diamine [Varlitinib)
  • the model was chosen due to high expression levels of HER proteins, and the combination of HER inhibition and DHODH inhibition was studied in the mouse studies.
  • mice Three groups of mice were dosed as follows:
  • Group 1 received lOOmg/Kg of Varlitinib BID (twice daily),
  • Group 2 received lOmg/Kg of ASLAN003 QD (once daily), and
  • Group 3 received lOOmg/Kg of Varlitinib BID and also lOmg/Kg of ASLAN003 QD.
  • Figures 1, 3 and 4 show immunoblots of tumours removed from mice after two days of treatment and show that combination therapy of varlitinib and ASLAN003 had increased pharmacodynamics effects even at after a short treatment period, as compared to varlitinib monotherapy.
  • Figure 5 shows combination of ASLAN003 with different dose levels of varlitinib and the combination therapy consistently shows increased tumour growth inhibition.
PCT/EP2016/070874 2015-09-04 2016-09-05 Combination of a dhodh inhibitor and a her inhibitor for use in the treatment of cancer WO2017037292A1 (en)

Applications Claiming Priority (16)

Application Number Priority Date Filing Date Title
GBGB1515718.3A GB201515718D0 (en) 2015-09-04 2015-09-04 Method
GBGB1515716.7A GB201515716D0 (en) 2015-09-04 2015-09-04 Method
GBGB1515712.6A GB201515712D0 (en) 2015-09-04 2015-09-04 Method
GB1515718.3 2015-09-04
GBGB1515714.2A GB201515714D0 (en) 2015-09-04 2015-09-04 Method
GB1515714.2 2015-09-04
GB1515716.7 2015-09-04
GB1515712.6 2015-09-04
US201562217346P 2015-09-11 2015-09-11
US201562217332P 2015-09-11 2015-09-11
US62/217,346 2015-09-11
US62/217,332 2015-09-11
GB1605583.2 2016-04-01
GB201605583 2016-04-01
GBGB1608660.5A GB201608660D0 (en) 2016-05-17 2016-05-17 Method
GB1608660.5 2016-05-17

Publications (1)

Publication Number Publication Date
WO2017037292A1 true WO2017037292A1 (en) 2017-03-09

Family

ID=58186741

Family Applications (4)

Application Number Title Priority Date Filing Date
PCT/EP2016/070874 WO2017037292A1 (en) 2015-09-04 2016-09-05 Combination of a dhodh inhibitor and a her inhibitor for use in the treatment of cancer
PCT/EP2016/070888 WO2017037298A1 (en) 2015-09-04 2016-09-05 A combination therapy comprising varlitinib and an anticancer agent
PCT/EP2016/070893 WO2017037300A1 (en) 2015-09-04 2016-09-05 Varlitinib for use in the treatment of resistant or refractory cancer
PCT/EP2016/070890 WO2017037299A1 (en) 2015-09-04 2016-09-05 Treatment of biliary duct cancer

Family Applications After (3)

Application Number Title Priority Date Filing Date
PCT/EP2016/070888 WO2017037298A1 (en) 2015-09-04 2016-09-05 A combination therapy comprising varlitinib and an anticancer agent
PCT/EP2016/070893 WO2017037300A1 (en) 2015-09-04 2016-09-05 Varlitinib for use in the treatment of resistant or refractory cancer
PCT/EP2016/070890 WO2017037299A1 (en) 2015-09-04 2016-09-05 Treatment of biliary duct cancer

Country Status (7)

Country Link
US (4) US10682353B2 (es)
EP (3) EP3344253B1 (es)
JP (3) JP2018526382A (es)
KR (2) KR20180043356A (es)
CN (4) CN108025010A (es)
ES (2) ES2788162T3 (es)
WO (4) WO2017037292A1 (es)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017184086A1 (en) * 2016-04-21 2017-10-26 Aslan Pharmaceuticals Pte. Ltd. Method of treating liver cancer
WO2019012030A1 (en) 2017-07-13 2019-01-17 INSERM (Institut National de la Santé et de la Recherche Médicale) DHODH INHIBITOR AND CHK1 INHIBITOR FOR THE TREATMENT OF CANCER
US10682353B2 (en) 2015-09-04 2020-06-16 Aslan Pharmaceuticals Pte Ltd Varlitinib for use in the treatment of resistant or refractory cancer
US11311548B2 (en) 2017-03-02 2022-04-26 Aslan Pharmaceuticals Pte. Ltd. Cancer therapy
US11382903B2 (en) 2017-03-02 2022-07-12 Aslan Pharmaceuticals Pte. Ltd. Cancer therapy

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019083455A1 (en) * 2017-10-25 2019-05-02 Aslan Pharmaceuticals Pte Ltd VARLITINIB FOR USE IN THE TREATMENT OF CANCER IN A PATIENT IDENTIFIED AS PRESENTING HER1, HER2 AND / OR HER3-ACTIVATED HERC-RECEPTOR CANCER CELLS
WO2019083457A1 (en) * 2017-10-25 2019-05-02 Aslan Pharmaceuticals Pte Ltd VARLITINIB FOR USE IN THE TREATMENT OF CANCER IN A PATIENT IDENTIFIED AS MUTATION OF THE BETA-CATENIN PATHWAY
WO2019083458A1 (en) * 2017-10-25 2019-05-02 Aslan Pharmaceuticals Pte Ltd VARLITINIB FOR THE TREATMENT OF CANCER TO REDUCE HYPOXIA
WO2019083456A1 (en) * 2017-10-25 2019-05-02 Aslan Pharmaceuticals Pte Ltd VARLITINIB FOR USE IN THE TREATMENT OF CANCER FOR NORMALIZING ANGIOGENESIS IN A CANCER MASS
WO2019199137A1 (ko) 2018-04-13 2019-10-17 주식회사 엘지화학 후처리 가교에 의한 분리막의 물성 향상 방법 및 이에 의한 분리막
EP3864014B1 (en) * 2018-10-09 2023-07-05 ASLAN Pharmaceuticals Pte Ltd Malonate salt of varlitinib
GB201913122D0 (en) 2019-09-11 2019-10-23 Seald As Compositions and methods for treatment of cholangiocarcinoma
GB201913123D0 (en) 2019-09-11 2019-10-23 Seald As Compositions and methods for treatment of cholangiocarcinoma
GB201913121D0 (en) 2019-09-11 2019-10-23 Seald As Compositions and methods for treatment of cholangiocarcinoma
GB201913124D0 (en) 2019-09-11 2019-10-23 Seald As Compositions and methods for treatment of cholangiocarcinoma

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005016346A1 (en) * 2003-08-14 2005-02-24 Array Biopharma Inc. Quinazoline analogs as receptor tyrosine kinase inhibitors
WO2012052179A1 (en) * 2010-10-22 2012-04-26 Almirall, S.A. Amino derivatives for the treatment of proliferative skin disorders

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PE20090486A1 (es) 2007-01-30 2009-04-27 Schering Corp Composiciones farmaceuticas y metodos para utilizar temozolomida e inhibidores de quinasa multidirigidos
GB0706633D0 (en) 2007-04-04 2007-05-16 Cyclacel Ltd Combination
WO2010127417A2 (en) 2009-05-05 2010-11-11 Katholieke Universiteit Leuven Hepatocellular carcinoma
AU2013211871B2 (en) 2012-01-25 2017-12-14 Board Of Regents, The University Of Texas System Biomarkers and combination therapies using oncolytic virus and immunomodulation
CA2877414A1 (en) 2012-07-30 2014-02-06 Alex Wah Hin Yeung Live and in-vivo tumor specified cancer vaccine system
ES2703208T3 (es) 2013-02-27 2019-03-07 Daiichi Sankyo Co Ltd Método de predicción de la sensibilidad a un compuesto que inhibe la vía de transducción de señales de MAPK
CN104415335A (zh) 2013-09-02 2015-03-18 北京中康万达医药科技有限公司 体内个体化系统免疫治疗方法和装置
US10350275B2 (en) 2013-09-21 2019-07-16 Advantagene, Inc. Methods of cytotoxic gene therapy to treat tumors
MX2016005283A (es) 2013-10-25 2017-02-20 Pharmacyclics Llc Tratamiento que utiliza inhibidores de tirosina quinasa de bruton e inmunoterapia.
CN106132439A (zh) 2014-03-31 2016-11-16 豪夫迈·罗氏有限公司 包含抗血管发生剂和ox40结合激动剂的组合疗法
AU2015335607B2 (en) 2014-10-24 2020-04-23 Calidi Biotherapeutics, Inc. Combination immunotherapy approach for treatment of cancer
US20190117655A1 (en) 2015-09-04 2019-04-25 Aslan Pharmaceuticals Pte Ltd Treatment of Biliary Duct Cancer
EP3344253B1 (en) 2015-09-04 2020-02-26 Aslan Pharmaceuticals PTE Limited Varlitinib for use in the treatment of resistant or refractory cancer
WO2017184086A1 (en) 2016-04-21 2017-10-26 Aslan Pharmaceuticals Pte. Ltd. Method of treating liver cancer
US10526294B2 (en) 2016-06-24 2020-01-07 Mersana Therapeutics, Inc. Pyrrolobenzodiazepines and conjugates thereof
GB201611580D0 (en) 2016-07-01 2016-08-17 Aslan Pharmaceuticals Pte Ltd Method
WO2019083455A1 (en) 2017-10-25 2019-05-02 Aslan Pharmaceuticals Pte Ltd VARLITINIB FOR USE IN THE TREATMENT OF CANCER IN A PATIENT IDENTIFIED AS PRESENTING HER1, HER2 AND / OR HER3-ACTIVATED HERC-RECEPTOR CANCER CELLS
WO2019083457A1 (en) 2017-10-25 2019-05-02 Aslan Pharmaceuticals Pte Ltd VARLITINIB FOR USE IN THE TREATMENT OF CANCER IN A PATIENT IDENTIFIED AS MUTATION OF THE BETA-CATENIN PATHWAY
WO2019083458A1 (en) 2017-10-25 2019-05-02 Aslan Pharmaceuticals Pte Ltd VARLITINIB FOR THE TREATMENT OF CANCER TO REDUCE HYPOXIA
WO2019083456A1 (en) 2017-10-25 2019-05-02 Aslan Pharmaceuticals Pte Ltd VARLITINIB FOR USE IN THE TREATMENT OF CANCER FOR NORMALIZING ANGIOGENESIS IN A CANCER MASS

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005016346A1 (en) * 2003-08-14 2005-02-24 Array Biopharma Inc. Quinazoline analogs as receptor tyrosine kinase inhibitors
WO2012052179A1 (en) * 2010-10-22 2012-04-26 Almirall, S.A. Amino derivatives for the treatment of proliferative skin disorders

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
DATABASE PubChem Compound [online] 20 July 2009 (2009-07-20), "Varlitinib", XP002764106, retrieved from NCBI Database accession no. 42642648 *
KIM J. ET AL.: "664P:Phase IIa study to evaluate the biological activity of ASLAN001 in HER-1/2 co-expressing or HER-2 amplified advanced gastric cancer", ANNALS OF ONCOLOGY, vol. 25, no. Suppl 4, 2014, pages iv226, XP002764105, DOI: 10.1093/annonc/mdu334.49 *
SUSAN ELLARD, ET AL.: "Abstract#3603: ARRY-334543 in ErbB2 positive metastatic breast cancer and other ErbB expressing-cancers: experience from expansion cohorts on a phase I study", CANCER RESEARCH, 18 April 2009 (2009-04-18) - 22 April 2009 (2009-04-22), 100th AACR Annual Meeting-- Apr 18-22, 2009; Denver, CO American Association for Cancer Research, XP002764103, ISSN: 0197-016X, Retrieved from the Internet <URL:http://cancerres.aacrjournals.org/content/69/9_Supplement/3603> [retrieved on 20161107] *
WANG DE-SHEN ET AL: "ARRY-334543 Reverses Multidrug Resistance by Antagonizing the Activity of ATP-Binding Cassette Subfamily G Member 2", JOURNAL OF CELLULAR BIOCHEMISTRY, vol. 115, no. 8, August 2014 (2014-08-01), pages 1381 - 1391, XP002764104 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10682353B2 (en) 2015-09-04 2020-06-16 Aslan Pharmaceuticals Pte Ltd Varlitinib for use in the treatment of resistant or refractory cancer
US10849899B2 (en) 2015-09-04 2020-12-01 Aslan Pharmaceuticals Pte Ltd Combination therapy comprising Varlitinib and an anticancer agent
WO2017184086A1 (en) * 2016-04-21 2017-10-26 Aslan Pharmaceuticals Pte. Ltd. Method of treating liver cancer
US11311548B2 (en) 2017-03-02 2022-04-26 Aslan Pharmaceuticals Pte. Ltd. Cancer therapy
US11382903B2 (en) 2017-03-02 2022-07-12 Aslan Pharmaceuticals Pte. Ltd. Cancer therapy
WO2019012030A1 (en) 2017-07-13 2019-01-17 INSERM (Institut National de la Santé et de la Recherche Médicale) DHODH INHIBITOR AND CHK1 INHIBITOR FOR THE TREATMENT OF CANCER

Also Published As

Publication number Publication date
JP2018526381A (ja) 2018-09-13
KR20180043356A (ko) 2018-04-27
US10357494B2 (en) 2019-07-23
CN108135901A (zh) 2018-06-08
KR20180048804A (ko) 2018-05-10
EP3344253A1 (en) 2018-07-11
US20180256578A1 (en) 2018-09-13
WO2017037300A1 (en) 2017-03-09
WO2017037299A1 (en) 2017-03-09
EP3344251B1 (en) 2021-04-28
EP3344252B1 (en) 2020-02-26
CN113750238A (zh) 2021-12-07
JP7332293B2 (ja) 2023-08-23
ES2788162T3 (es) 2020-10-20
US20200009144A1 (en) 2020-01-09
CN108025010A (zh) 2018-05-11
US20180353510A1 (en) 2018-12-13
US10849899B2 (en) 2020-12-01
JP2018526382A (ja) 2018-09-13
CN108135900A (zh) 2018-06-08
ES2882031T3 (es) 2021-12-01
EP3344253B1 (en) 2020-02-26
US10682353B2 (en) 2020-06-16
EP3344252A1 (en) 2018-07-11
EP3344251A1 (en) 2018-07-11
US20180243302A1 (en) 2018-08-30
JP2018535923A (ja) 2018-12-06
WO2017037298A1 (en) 2017-03-09

Similar Documents

Publication Publication Date Title
WO2017037292A1 (en) Combination of a dhodh inhibitor and a her inhibitor for use in the treatment of cancer
RU2757373C2 (ru) Комбинированная терапия противоопухолевым алкалоидом
WO2018136009A1 (en) Combination therapy
WO2018136010A1 (en) Combination therapy
US20190134034A1 (en) Method of Treating Liver Cancer
JP2012515184A (ja) 大腸がんの治療方法
WO2010086964A1 (ja) がん治療のための併用療法
WO2017200492A1 (en) Method of treating cancer
WO2018222134A1 (en) Cancer therapy
KR20070089158A (ko) 암의 치료를 위한n-(3-메톡시-5-메틸피라진-2-일)-2-(4-[1,3,4-옥사디아졸-2-일]페닐)피리딘-3-설폰아미드 및 항-유사분열제의 조합물
WO2018222135A1 (en) Cancer therapy
AU2017288094A1 (en) Maintenance therapy for the treatment of cancer
US20190117655A1 (en) Treatment of Biliary Duct Cancer
WO2017081676A1 (en) Pharmaceutical compositions and methods for treatment of cancer
JP2022543021A (ja) がんを処置する方法及び使用
TW201904570A (zh) 治療肝癌之方法
TW201313225A (zh) 與放射線治療關聯之包含奧瑞布林(ombrabulin)及順鉑(cisplatin)之抗腫瘤組合

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 16774457

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 18/06/2018)

122 Ep: pct application non-entry in european phase

Ref document number: 16774457

Country of ref document: EP

Kind code of ref document: A1